Resveratrol inhibits hypoxia-driven ROS-induced invasive and migratory ability of pancreatic cancer cells via suppression of the Hedgehog signaling pathway

  • Authors:
    • Wei Li
    • Lei Cao
    • Xin Chen
    • Jianjun Lei
    • Qingyong Ma
  • View Affiliations

  • Published online on: December 22, 2015     https://doi.org/10.3892/or.2015.4504
  • Pages: 1718-1726
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A hypoxic microenvironment is commonly found in the central region of solid tumors, including pancreatic cancer. Our previous study revealed that resveratrol plays an important role in suppressing the proliferation and EMT of pancreatic cancer cells. However, whether resveratrol could suppress hypoxia-induced cancer progression and the underlying mechanisms have not been fully elucidated. The aim of the present study was to evaluate whether resveratrol affects hypoxia-induced reactive oxygen species (ROS) production and the activation of the Hedgehog (Hh) signaling pathway as well as the invasion of pancreatic cancer. The human pancreatic cancer cell lines, BxPC-3 and Panc-1, were subjected to a hypoxic condition and three different concentrations of resveratrol. The intracellular ROS were determined using 2,7-dichlorodihydrofluorecein diacetate. Wound healing and Transwell invasion assays were used to detect the migratory and invasive potential of the cancer cells. Metastatic-related and Hh signaling-related factors were detected by qRT-PCR and western blot analysis. Immunofluorescence staining was used to test the nuclear translocation of GLI1. The results showed that the hypoxia-induced production of ROS was decreased by resveratrol in a concentration-dependent manner. Resveratrol significantly inhibited the hypoxia-stimulated invasion and migration of pancreatic cancer cells. Resveratrol inhibited hypoxia-induced HIF-1α protein expression. Resveratrol also suppressed hypoxia‑induced expression of metastatic-related factors, uPA and MMP2. In addition, resveratrol markedly inhibited hypoxia-mediated activation of the Hh signaling pathway. Furthermore, the antioxidant N-acetylcysteine (NAC) significantly suppressed the invasive and migratory ability of pancreatic cancer cells during hypoxia. Taken together, these data indicate that resveratrol plays an important role in suppressing hypoxia-driven ROS-induced pancreatic cancer progression by inhibiting the Hh signaling pathway, providing evidence that resveratrol may be a potential candidate for the chemoprevention of cancer.

Introduction

Pancreatic cancer is a highly malignant tumor with an extremely poor prognosis, partially due to the lack of early diagnosis and treatment options (1). Although surgery remains the only way to cure this severe disease, the majority of patients present at an advanced inoperable stage and only 20% of patients are with localized disease amenable for surgery (2). Even those seemingly resectable pancreatic tumors often fail to be cured due to the microscopic systemic spread of the cancer that occurs before the surgical intervention (3). Understanding the molecular basis of the disease is highly desirable for developing new strategies to prevent and treat pancreatic cancer.

Low oxygen tension is most commonly presented in the microenvironment of solid tumors (4). Tumor hypoxia is associated with enhanced tumor invasiveness, angiogenesis and distant metastasis (5,6). Hypoxia-inducible factor-1 (HIF-1), which belongs to the basic helix-loop-helix-periodic acid-Schiff domain transcription factor family, is the most important transcription factor as a result of intratumoral hypoxia (7). HIF-1 consists of two subunits, HIF-1α and HIF-1β. Only the expression and activation of HIF-1α is tightly regulated by the cellular oxygen concentration (8). In pancreatic cancer, the level of HIF-1α expression is overexpressed and is associated with tumor progression, angiogenesis, cell migration and hepatic metastasis (9,10). Our previous study identified that Hedgehog (Hh) signaling modulated hypoxia-induced pancreatic cancer epithelial to mesenchymal transition (EMT) and invasion (11).

The Hh signaling pathway, which is considered to play an important role in vertebrate development, the homeostatic process and tumorigenesis (12), is normally quiescent in the adult pancreas and has been shown to be very active in pancreatic cancer (11). The Hh signaling pathway, initiated through the binding of secreted Hh ligands to the membrane receptor patched 1 (PTCH1), results in smoothened (SMO) dissociation, nuclear translocation and activation of the transcription factors of the GLI family (11,13). The expression of SMO and GLI1 is presumed to be markers of Hh pathway activation (11). Our previous study confirmed that hypoxia-induced invasion and the EMT process is intimately related with the Hh signaling pathway (11). In addition, inhibition of Hh signaling also enhanced vascular density and delivery of gemcitabine in a mouse model of pancreatic cancer (14).

Reactive oxygen species (ROS) generated by the mitochondrial respiratory chain, consist of a number of chemically reactive molecules derived from oxygen, including hydrogen peroxide (H2O2). Malignant tumor cells commonly have increased levels of ROS, which plays a significant role in cancer progression (15,16). Our recent study showed that hypoxia-induced ROS production is intimately related with pancreatic stellate cell (PSC) activation and pancreatic cancer cell invasion (17).

Resveratrol (trans-3,4′,5-trihydroxystilbene), a natural polyphenolic phytoalexin, is widely found in plants (such as grape skin, red wine, berries and peanuts) and in traditional Chinese medicines (such as Rheum officinale Baill. and Polygonum cuspidatum) (18). Recent studies have shown that resveratrol has many biological and pharmaceutical properties, including anti-inflammatory, antioxidant, anti-aging, neuroprotective and antitumorigenic capabilities (1921). Our previous study demonstrated that resveratrol plays an important role in suppressing the proliferation and EMT of pancreatic cancer cells via the PI-3K/Akt/NF-κB signaling pathway (22). In addition, we also confirmed that resveratrol inhibited the growth of human pancreatic cancer cells in vitro by inhibiting cell proliferation and promoting cell apoptosis via inhibition of the Hh signaling pathway (23).

In the present study, we tested the hypothesis that resveratrol is able to inhibit hypoxia-induced ROS production and the invasive and migratory ability of pancreatic cancer cells. We also investigated the effect of resveratrol on hypoxia-induced activation of the Hh pathway. Results from the present study suggest that resveratrol treatment may be a novel option for the therapy of pancreatic cancer via inhibition of the Hh signaling pathway.

Materials and methods

Preparation of chemicals

Dulbecco's modified eagle's medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco (Grand Island, NY, USA). Resveratrol (>99% pure) was acquired from Xi'an Chongxin Natural Additive Company (Xi'an, China). N-acetylcysteine (NAC) was purchased from Sigma. Millicell Transwells for the invasion assays were obtained from Millipore (Billerica, MA, USA). Matrigel was from BD (Biosciences, Bedford, MA, USA). Primary antibodies against HIF-1α, MMP-2, uPA, SHH, SMO as well as GLI1 were from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Nitrocellulose membranes were from Millipore. The BCA assay kit and the chemiluminescence kit were from Pierce (Rockford, IL, USA). Other reagents were purchased from common commercial sources. All drug solutions were freshly prepared on the day of testing.

Cell cultures and treatments

The human pancreatic cancer cell lines, BxPC-3 and Panc-1, were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were cultured in DMEM containing 10% dialyzed heat-inactivated FBS, 100 U/ml penicillin and 100 μg/ml streptomycin in a humidified atmosphere of 5% CO2 at 37°C. In experiments designed to assess the role of hypoxia, cells were first cultured in normoxic conditions to obtain the desired subconfluence level (65–70%), and were then incubated in strictly controlled hypoxic conditions (1% O2). Exponentially growing cells in complete medium were pretreated for 1 h with different concentrations of resveratrol, followed by continual incubation in normal culturing conditions or hypoxic conditions for the indicated time intervals according to the aim of the experiment.

Measurement of intracellular ROS

The level of intracellular ROS was measured using the ROS assay kit. In brief, cells were incubated with 2,7-dichlorodihydrofluorecein diacetate (DCFDA) for 30 min, washed in phosphate-buffered saline (PBS) 3 times, and fluorescence intensity was measured using a fluorometer (Becton-Dickinson, USA) with excitation at 488 nm and emission at 525 nm.

Wound healing assay

Cell migratory ability was detected by a wound-healing assay. Pancreatic cancer cells were seeded into 24-well plates (1.0×105 cells/500 μl). After the cells grew to 90–100% confluency, a sterile pipette tip was used to produce a wound line between the cells. Cellular debris was removed by washing with PBS and then allowed to migrate for 24 h. Images were captured at time 0 and 24 h post-wounding under a Nikon Diaphot TMD inverted microscope (magnification, ×10). The relative distance traveled by the leading edge from 0 to 24 h was assessed using Photoshop software (n=5).

Transwell Matrigel invasion assays

The invasion of pancreatic cancer cells was performed in Transwell chambers. The 8.0-μm pore inserts were coated with 25 μl Matrigel. The cell suspensions (5×104) were added to the upper chambers in DMEM containing 1% FBS. Simultaneously, 500 ml of DMEM containing 20% FBS was placed in the lower chambers. The cells were allowed to migrate for 48 h at 37°C. The non-invading cells were removed from the upper surface by scraping with a wet cotton swab. After rinsing with PBS, the filter was fixed and stained with crystal violet. Invasion ability was determined by counting the stained cells. The invasion ability was determined by counting the stained cells on the bottom surface. Three random fields were captured at a magnification of ×20 (n=3).

Real-time quantitative PCR (qRT-PCR)

Total RNA was extracted from the pancreatic cancer cells using the Fastgen200 RNA isolation system (Fastgen, Shanghai, China) according to the manufacturer's protocol. Total RNA was reverse-transcribed into cDNA using a PrimeScript RT reagent kit (Takara, Dalian, China). The primer sequences were as follows: HIF-1α-F, 5′-AAG TCT AGG GAT GCA GCA-3′ and HIF-1α-R, 5′-CAA GAT CAC CAG CAT CAT G-3′; MMP-2-F, 5′-GAT GAT GCC TTT GCT CGT GC-3′ and MMP-2-R, 5′-CAA AGG GGT ATC CAT CGC CA-3′; uPA-F, 5′-TAA GAG CTG GTG TCT GAT TG-3′ and uPA-R, 5′-TTG GAT GAA CTA GGC TAA AA-3′; SHH-F, 5′-TCC AGA AAC TCC GAG CGA TTT AAG-3′ and SHH-R, 5′-CAC TTC CTG GCC ACT GGT TCA-3′; SMO-F, 5′-ACG AGG ACG TGG AGG GCT G-3′ and SMO-R, 5′-CGC ACG GTA TCG GTA GTT CT-3′; GLI1-F, 5′-GGG ATG ATC CCA CAT CCT CAG TC-3′ and GLI1-R, 5′-CTG GAG CAG CCC CCC CAG T-3′; β-actin-F, 5′-GAC TTA GTT GCG TTA CAC CCT TTC T-3′ and β-actin-R, 5′-GAA CGG TGA AGG TGA CAG CAG T-3′. The PCR reactions consisted of 30 sec at 95°C followed by 40 cycles at 95°C for 5 sec, at 60°C for 30 sec and at 72°C for 30 sec. After each qRT-PCR, a dissociation curve analysis was conducted. Relative gene expression was calculated using the 2−ΔΔCt method as previously reported (24).

Western blotting

Proteins were electrophoretically resolved on a denaturing SDS-polyacrylamide gel and electrotransferred onto nitrocellulose membranes. The membranes were initially blocked with 5% non-fat dry milk in Tris-buffered saline (TBS) for 2 h and then probed with antibodies against HIF-1α, MMP-2, uPA, SHH, SMO, GLI1 or β-actin (loading control). After co-incubation with the primary antibodies at 4°C overnight, the membranes were blotted with the secondary antibody for 2 h at 37°C. The results were visualized using the ECL western blotting substrate and photographed by GeneBox (SynGene).

Immunofluorescence microscopy

After the designated treatment, pancreatic cancer cells were fixed with 4% paraformaldehyde for 10 min at room temperature, permeabilized in 0.5% Triton X-100 for 10 min and blocked in 1% BSA for 1 h. Fixed cells were then incubated with primary antibody against GLI1 (1:100) at 4°C overnight. Cells were washed and incubated with fluorescein isothiocyanate (FITC)-conjugated goat anti-rabbit IgG for 1 h in a darkroom. Nuclei were stained with DAPI for 5 min. The cells were visualized by a fluorescence microscope (Nikon, Japan) using appropriate excitation and emission spectra at a magnification of ×400.

Statistical analysis

Statistical analysis was performed using SPSS software (version 17.0; SPSS, Inc., Chicago, IL, USA). Data are presented as the means ± SEM of three replicate assays. Differences between groups were analyzed by analysis of variance (ANOVA). Statistical significance was set at P<0.05. All experiments were independently repeated at least three times.

Results

Resveratrol decreases hypoxia-induced production of ROS in pancreatic cancer cells

The intracellular ROS levels in BxPC-3 and Panc-1 cells treated with different concentrations of resveratrol under hypoxia conditions were determined using cell-permeable and redox-sensitive compound DCFDA by flow cytometry. Our previous study confirmed that the 50% inhibitory concentration (IC50) for both BxPC-3 and Panc-1 cells is ~50 μM of resveratrol, which exhibits no cytotoxic effects on BxPC-3 and Panc-1 cells (22). Therefore, treatment concentrations of 12.5, 25 and 50 μM of resveratrol on the cells were used for the present experiments. As shown in Fig. 1, a hypoxia condition significantly increased intracellular levels of ROS, and resveratrol suppressed these effects in a concentration-dependent manner after incubation for 24 h.

Resveratrol suppresses hypoxia-induced invasive ability of pancreatic cancer cells

A vital step of cancer metastasis is invasion of cancer cells through the basement membrane. In order to confirm whether resveratrol influences hypoxia-induced cancer cell invasive ability, we used a Transwell invasion assay. As shown in Fig. 2, hypoxia exposure significantly increased pancreatic cancer invasive ability, while the average cell number that invaded into the lower chamber decreased as the resveratrol concentration increased from 12.5 to 50 μM.

Resveratrol inhibits hypoxia-induced wound closure of pancreatic cancer cells

Migration and invasion are two important aspects that lead to the ability of cancer cells to form metastases. The effect of resveratrol on hypoxia-induced pancreatic cancer cell motility was determined using a wound-healing assay. Results showed that a hypoxic condition caused a significant increase in the migration of both BxPC-3 and Panc-1 cells after incubation for 24 h. Resveratrol suppressed these effects in a dose-dependent manner (Fig. 3). This finding revealed that resveratrol may be an effective inhibitor of hypoxia-induced migration and invasion of pancreatic cancer cells.

Resveratrol inhibits the expression of hypoxia-induced HIF-1α and metastatic-related factors

Previous studies have demonstrated that the effect induced by hypoxia is mainly mediated by HIF-1α (25). In order to investigate the effect of hypoxia on pancreatic cancer cells, both BxPC-3 and Panc-1 cells were exposed to hypoxic conditions (1% O2) for up to 48 h. As shown in Fig. 4, the expression levels of HIF-1α, uPA and MMP-2 were markedly increased in both cell lines, compared with normoxic conditions. Resveratrol decreased hypoxia-induced HIF-1α protein expression (Fig. 4A), while no apparent changes in HIF-1α mRNA were observed in both the BxPC-3 and Panc-1 cells (Fig. 4B), which indicated that resveratrol inhibits hypoxia-induced HIF-1α protein expression through a post-transcriptional mechanism as metastatic-related factors, uPA and MMP-2, have been implicated in cancer invasion and metastasis. Our results showed that resveratrol suppressed hypoxia-induced uPA and MMP-2 expression at both the mRNA and protein levels (Fig. 4).

Resveratrol downregulates the hypoxia-activated Hh signaling pathway

Hh signaling plays an important role in the initiation and progression of pancreatic cancer (26). As shown in Fig. 4, the mRNA and protein expression levels of SHH, SMO and GLI1 were significantly increased in both the BxPC-3 and Panc-1 cancer cells, compared with the normal controls, which indicated that Hh signaling was activated in both cell lines under hypoxic condition. Resveratrol markedly decreased hypoxia-induced expression levels of SHH, SMO and GLI1. In addition, immunofluorescence staining of these treated cells also confirmed that a hypoxic condition could induce GLI1 expression in the nucleus of BxPC-3 and Panc-1 cells, while resveratrol obviously decreased the nuclear translocation of GLI1 (Fig. 5).

NAC suppresses hypoxia-induced ROS generation as well as the invasive and migratory ability of pancreatic cancer cells

NAC, a precursor of L-cysteine, is thought to be a scavenger of free radicals such as hydroxyl radical, H2O2 and superoxide. To explore whether hypoxia-induced invasive and migratory ability of pancreatic cancer cells is related with ROS production, we cultured the cells under a hypoxic condition in the presence or absence of 20 mM NAC. The results showed that NAC efficiently reduced ROS levels under a hypoxic condition in both the BxPC-3 and Panc-1 cells (Fig. 6A). The average cell number that invaded into the lower chamber decreased with NAC treatment (Fig. 6B). The cell migration ability (Fig. 6C) was also significantly inhibited 24 h after the addition of NAC. Additionally, the mRNA expression of MMP-2, uPA and GLI1 was downregulated by NAC under a hypoxic condition (Fig. 6D). Taken together, our results demonstrated that resveratrol inhibits hypoxia-driven ROS-induced cancer progression via suppression of the Hh signaling pathway in both the BxPC-3 and Panc-1 cells.

Discussion

Pancreatic cancer is a malignant carcinoma of the digestive system with an extremely high mortality rate, due to both the inherently aggressive biology of the disease and its late diagnosis in most cases (2). A hypoxic microenvironment is commonly found in the central region of solid tumors, including pancreatic cancer (11). Tumor hypoxia not only increases the metastatic capacity of cancer cells, but also leads to resistance to chemotherapy and radiotherapy. Hypoxia can also induce altered transcription and translation of a number of DNA damage response and repair genes, which further leads to inhibition of recombination-mediated repair of DNA double-strand breaks. In addition, hypoxia can increase the rate of mutation (27). Overexpression of HIF-1α has been shown in many human cancers and their metastases and is closely associated with a more aggressive tumor progression (28). Our previous study demonstrated that resveratrol inhibits the growth of human pancreatic cancer cells in vitro by inhibiting cell proliferation and promoting cell apoptosis via inhibition of the Hh signaling pathway (23). In the present study, we focused on whether resveratrol is able to suppress hypoxia-induced cancer invasive and migratory ability and its underlying mechanism.

Our data showed that a hypoxic condition could significantly increase the production of ROS and the expression of HIF-1α as well as cancer metastatic-related factors, uPA and MMP-2, in BxPC-3 and Panc-1 cells, which further enhanced the capacity of the pancreatic cancer cells to migrate and invade the extracellular matrix. Resveratrol was able to abrogate these effects of a hypoxic condition. A previous study confirmed that hypoxia activates canonical Hh signaling through accumulation of HIF-1α (29). In the present study, we tested the effects of a hypoxic condition and resveratrol on the activation of SHH, SMO and GLI1. The data showed that a hypoxic condition significantly increased the expression levels of SHH, SMO and GLI1 in both the BxPC-3 and Panc-1 cancer cells, whereas the addition of resveratrol to the cell culture resulted in a decrease in these Hh pathway-related factors. In addition, the hypoxia-enhanced nuclear translocation of GLI1 was decreased by resveratrol.

ROS generated by the mitochondrial respiratory chain, consist of a number of chemically reactive molecules derived from oxygen. As a double-edged sword, excess ROS production can kill cancer cells, whereas sublethal concentrations of ROS can stimulate tumor progression by promoting cell proliferation, survival, invasion and metastasis (30). Our previous studies confirmed that both a hyperglycemic condition and SOD-induced ROS production were able to promote the invasive and migratory activity of pancreatic cancer (15,16). Our recent study also showed that hypoxia-induced ROS production is intimately related with pancreatic stellate cell (PSC) activation and pancreatic cancer cell invasion (17). In the present study, we found that hypoxia-induced ROS production was suppressed by resveratrol in a concentration-dependent manner.

Resveratrol and its analogues have been proven to inhibit the invasion of many tumor types, including pancreatic cancer (31). Recent studies have focused on the relationship between resveratrol and hypoxia-induced cancer progression. Wu et al (8) demonstrated that the anti-metastatic effect of resveratrol was associated with the restriction of invasion, mobility, adhesion and MMP expression under both normoxic and hypoxic conditions in colon carcinoma. Mitani et al (32) showed that dietary resveratrol inhibited β-catenin-mediated androgen receptor function by decreasing the expression of HIF-1α protein in hypoxic LNCaP cells and consequently suppressed prostate cancer cell growth in vivo. They also confirmed that resveratrol suppressed hypoxia-induced resistance to cytotoxicity of doxorubicin and repressed the expression of CBR1 in breast cancer cells (33). In the present study, we also found that resveratrol was able to decrease hypoxia-induced pancreatic cancer invasion and migration, which may be attributed to the reduction of ROS.

Resveratrol can inhibit tumor biological behavior through multiple signaling pathways. Our previous study indicated that resveratrol plays an important role in suppressing the proliferation, migration and invasion of pancreatic cancer cells in vitro by modulating EMT-related factors via the PI-3K/Akt/NF-κB signaling pathway. We also showed that resveratrol was able to suppress the migration and invasion of pancreatic cancer cells by inhibiting TGF-β-mediated EMT (22). Ji et al (34) confirmed that resveratrol downregulated MALAT1 and decreased nuclear localization of β-catenin, which in turn attenuated the Wnt/β-catenin signaling pathway leading to the inhibition of invasion and metastasis of colorectal cancer cells. Sun et al (35) showed that resveratrol activated SIRT1, which further hampered lung cancer cell metastasis in vivo. In addition, resveratrol also inhibited hypoxia-induced HIF-1α accumulation and vascular endothelial growth factor (VEGF) expression in both human tongue squamous cell carcinomas and hepatoma cells via the suppression of ERK1/2 and Akt signaling pathway (36).

Hh signaling activation is a common event in pancreatic cancer. The Hh signaling pathway is composed of patched (PTCH), a 12-transmembrane receptor, smoothened (SMO), a 7-transmembrane receptor and the GLI transcription factor family. A previous study demonstrated that hypoxia could activate canonical Hh signaling through accumulation of HIF-1α in vitro and in vivo (26). Our recent study also confirmed that accumulated HIF-1α could also trigger non-canonical Hh signaling to facilitate hypoxia-induced EMT and invasion processes in pancreatic cancer (11). Qin et al (23) recently indicated that resveratrol inhibited pancreatic cancer cell proliferation and promoted cell apoptosis via inhibition of the Hh signaling pathway. Gao et al (37) also demonstrated that resveratrol was able to inhibit gastric cancer cell invasion and metastasis in vitro by inhibiting the Hh signaling pathway and EMT. In addition, resveratrol could not only effectively downregulate interleukin-6-stimulated SHH signaling in human acute myeloid leukemia (38), but also inhibited both SHH signaling and Bcr-Abl expression in human chronic myeloid leukemia cells (39), indicating that resveratrol may have potential as a treatment for myeloid leukemia. In the present study, we showed that resveratrol was able to suppress hypoxia-induced activation of the Hh pathway and thus inhibited pancreatic cancer cell invasive and migratory ability.

In conclusion, the present study demonstrated that resveratrol plays an important role in suppressing hypoxia-induced migration and invasion of pancreatic cancer cells in vitro by inhibiting the Hh signaling pathway. These results suggest that resveratrol may be a potential anticancer agent for the treatment of pancreatic cancer.

Acknowledgments

The present study was supported by a grant from the National Natural Science Foundation of China (grant serial no. 81301846).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Castellanos EH, Cardin DB and Berlin JD: Treatment of early-stage pancreatic cancer. Oncology. 25:182–189. 2011.PubMed/NCBI

4 

Semenza GL: Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci. 33:207–214. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Hoffmann AC, Mori R, Vallbohmer D, Brabender J, Klein E, Drebber U, Baldus SE, Cooc J, Azuma M, Metzger R, et al: High expression of HIF1α is a predictor of clinical outcome in patients with pancreatic ductal adenocarcinomas and correlated to PDGFA, VEGF, and bFGF. Neoplasia. 10:674–679. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Zhao T, Gao S, Wang X, Liu J, Duan Y, Yuan Z, Sheng J, Li S, Wang F, Yu M, et al: Hypoxia-inducible factor-1α regulates chemotactic migration of pancreatic ductal adenocarcinoma cells through directly transactivating the CX3CR1 gene. PLoS One. 7:e433992012. View Article : Google Scholar

7 

Pouysségur J, Dayan F and Mazure NM: Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature. 441:437–443. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Wu H, Liang X, Fang Y, Qin X, Zhang Y and Liu J: Resveratrol inhibits hypoxia-induced metastasis potential enhancement by restricting hypoxia-induced factor-1 alpha expression in colon carcinoma cells. Biomed Pharmacother. 62:613–621. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Matsuo Y, Ding Q, Desaki R, Maemura K, Mataki Y, Shinchi H, Natsugoe S and Takao S: Hypoxia inducible factor-1 alpha plays a pivotal role in hepatic metastasis of pancreatic cancer: An immunohistochemical study. J Hepatobiliary Pancreat Sci. 21:105–112. 2014. View Article : Google Scholar

10 

Zhao X, Gao S, Ren H, Sun W, Zhang H, Sun J, Yang S and Hao J: Hypoxia-inducible factor-1 promotes pancreatic ductal adenocarcinoma invasion and metastasis by activating transcription of the actin-bundling protein fascin. Cancer Res. 74:2455–2464. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Lei J, Ma J, Ma Q, Li X, Liu H, Xu Q, Duan W, Sun Q, Xu J, Wu Z, et al: Hedgehog signaling regulates hypoxia induced epithelial to mesenchymal transition and invasion in pancreatic cancer cells via a ligand-independent manner. Mol Cancer. 12:662013. View Article : Google Scholar : PubMed/NCBI

12 

McMahon AP, Ingham PW and Tabin CJ: Developmental roles and clinical significance of hedgehog signaling. Curr Top Dev Biol. 53:1–114. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Nguyen NT, Lin DP, Yen SY, Tseng JK, Chuang JF, Chen BY, Lin TA, Chang HH and Ju JC: Sonic hedgehog promotes porcine oocyte maturation and early embryo development. Reprod Fertil Dev. 21:805–815. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, et al: Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 324:1457–1461. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Li W, Cao L, Han L, Xu Q and Ma Q: Superoxide dismutase promotes the epithelial-mesenchymal transition of pancreatic cancer cells via activation of the H2O2/ERK/NF-κB axis. Int J Oncol. 46:2613–2620. 2015.

16 

Li W, Ma Q, Li J, Guo K, Liu H, Han L and Ma G: Hyperglycemia enhances the invasive and migratory activity of pancreatic cancer cells via hydrogen peroxide. Oncol Rep. 25:1279–1287. 2011.PubMed/NCBI

17 

Lei J, Huo X, Duan W, Xu Q, Li R, Ma J, Li X, Han L, Li W, Sun H, et al: α-Mangostin inhibits hypoxia-driven ROS-induced PSC activation and pancreatic cancer cell invasion. Cancer Lett. 347:129–138. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Chen BY, Kuo CH, Liu YC, Ye LY, Chen JH and Shieh CJ: Ultrasonic-assisted extraction of the botanical dietary supplement resveratrol and other constituents of Polygonum cuspidatum. J Nat Prod. 75:1810–1813. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Albani D, Polito L, Signorini A and Forloni G: Neuroprotective properties of resveratrol in different neurodegenerative disorders. Biofactors. 36:370–376. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Fulda S: Resveratrol and derivatives for the prevention and treatment of cancer. Drug Discov Today. 15:757–765. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Jha RK, Ma Q, Sha H and Palikhe M: Emerging role of resveratrol in the treatment of severe acute pancreatitis. Front Biosci. 2:168–175. 2010. View Article : Google Scholar

22 

Li W, Ma J, Ma Q, Li B, Han L, Liu J, Xu Q, Duan W, Yu S, Wang F, et al: Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-κB pathway. Curr Med Chem. 20:4185–4194. 2013. View Article : Google Scholar

23 

Qin Y, Ma Z, Dang X, Li W and Ma Q: Effect of resveratrol on proliferation and apoptosis of human pancreatic cancer MIA PaCa-2 cells may involve inhibition of the Hedgehog signaling pathway. Mol Med Rep. 10:2563–2567. 2014.PubMed/NCBI

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔC T method. Methods. 25:402–408. 2001. View Article : Google Scholar

25 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Spivak-Kroizman TR, Hostetter G, Posner R, Aziz M, Hu C, Demeure MJ, Von Hoff D, Hingorani SR, Palculict TB, Izzo J, et al: Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer. Cancer Res. 73:3235–3247. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Luoto KR, Kumareswaran R and Bristow RG: Tumor hypoxia as a driving force in genetic instability. Genome Integr. 4:52013. View Article : Google Scholar : PubMed/NCBI

28 

Ye LY, Zhang Q, Bai XL, Pankaj P, Hu QD and Liang TB: Hypoxia-inducible factor 1α expression and its clinical significance in pancreatic cancer: A meta-analysis. Pancreatology. 14:391–397. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Bijlsma MF, Groot AP, Oduro JP, Franken RJ, Schoenmakers SH, Peppelenbosch MP and Spek CA: Hypoxia induces a hedgehog response mediated by HIF-1alpha. J Cell Mol Med. 13:2053–2060. 2009. View Article : Google Scholar

30 

Nishikawa M, Hashida M and Takakura Y: Catalase delivery for inhibiting ROS-mediated tissue injury and tumor metastasis. Adv Drug Deliv Rev. 61:319–326. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Xu Q, Zong L, Chen X, Jiang Z, Nan L, Li J, Duan W, Lei J, Zhang L, Ma J, et al: Resveratrol in the treatment of pancreatic cancer. Ann NY Acad Sci. 1348:10–19. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Mitani T, Harada N, Tanimori S, Nakano Y, Inui H and Yamaji R: Resveratrol inhibits hypoxia-inducible factor-1α-mediated androgen receptor signaling and represses tumor progression in castration-resistant prostate cancer. J Nutr Sci Vitaminol. 60:276–282. 2014. View Article : Google Scholar

33 

Mitani T, Ito Y, Harada N, Nakano Y, Inui H, Ashida H and Yamaji R: Resveratrol reduces the hypoxia-induced resistance to doxorubicin in breast cancer cells. J Nutr Sci Vitaminol. 60:122–128. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Ji Q, Liu X, Fu X, Zhang L, Sui H, Zhou L, Sun J, Cai J, Qin J, Ren J, et al: Resveratrol inhibits invasion and metastasis of colorectal cancer cells via MALAT1 mediated Wnt/β-catenin signal pathway. PLoS One. 8:e787002013. View Article : Google Scholar

35 

Sun L, Li H, Chen J, Dehennaut V, Zhao Y, Yang Y, Iwasaki Y, Kahn-Perles B, Leprince D, Chen Q, et al: A SUMOylation-dependent pathway regulates SIRT1 transcription and lung cancer metastasis. J Natl Cancer Inst. 105:887–898. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Zhang Q, Tang X, Lu QY, Zhang ZF, Brown J and Le AD: Resveratrol inhibits hypoxia-induced accumulation of hypoxia-inducible factor-1alpha and VEGF expression in human tongue squamous cell carcinoma and hepatoma cells. Mol Cancer Ther. 4:1465–1474. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Gao Q, Yuan Y, Gan HZ and Peng Q: Resveratrol inhibits the hedgehog signaling pathway and epithelial-mesenchymal transition and suppresses gastric cancer invasion and metastasis. Oncol Lett. 9:2381–2387. 2015.PubMed/NCBI

38 

Su YC, Li SC, Wu YC, Wang LM, Chao KS and Liao HF: Resveratrol downregulates interleukin-6-stimulated sonic hedgehog signaling in human acute myeloid leukemia. Evid Based Complement Alternat Med. 2013:5474302013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 35 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li W, Cao L, Chen X, Lei J and Ma Q: Resveratrol inhibits hypoxia-driven ROS-induced invasive and migratory ability of pancreatic cancer cells via suppression of the Hedgehog signaling pathway. Oncol Rep 35: 1718-1726, 2016
APA
Li, W., Cao, L., Chen, X., Lei, J., & Ma, Q. (2016). Resveratrol inhibits hypoxia-driven ROS-induced invasive and migratory ability of pancreatic cancer cells via suppression of the Hedgehog signaling pathway. Oncology Reports, 35, 1718-1726. https://doi.org/10.3892/or.2015.4504
MLA
Li, W., Cao, L., Chen, X., Lei, J., Ma, Q."Resveratrol inhibits hypoxia-driven ROS-induced invasive and migratory ability of pancreatic cancer cells via suppression of the Hedgehog signaling pathway". Oncology Reports 35.3 (2016): 1718-1726.
Chicago
Li, W., Cao, L., Chen, X., Lei, J., Ma, Q."Resveratrol inhibits hypoxia-driven ROS-induced invasive and migratory ability of pancreatic cancer cells via suppression of the Hedgehog signaling pathway". Oncology Reports 35, no. 3 (2016): 1718-1726. https://doi.org/10.3892/or.2015.4504